@article{anderson_tsvetkov_galante_devries_mccue_wood_barry_berto_lavin_taniguchi_et al._2023, title={Epigenetic function during heroin self-administration controls future relapse-associated behavior in a cell type-specific manner}, volume={120}, url={http://dx.doi.org/10.1073/pnas.2210953120}, DOI={10.1073/pnas.2210953120}, abstractNote={Opioid use produces enduring associations between drug reinforcement/euphoria and discreet or diffuse cues in the drug-taking environment. These powerful associations can trigger relapse in individuals recovering from opioid use disorder (OUD). Here, we sought to determine whether the epigenetic enzyme, histone deacetylase 5 (HDAC5), regulates relapse-associated behavior in an animal model of OUD. We examined the effects of nucleus accumbens (NAc) HDAC5 on both heroin- and sucrose-seeking behaviors using operant self-administration paradigms. We utilized cre-dependent viral-mediated approaches to investigate the cell-type–specific effects of HDAC5 on heroin-seeking behavior, gene expression, and medium spiny neuron (MSN) cell and synaptic physiology. We found that NAc HDAC5 functions during the acquisition phase of heroin self-administration to limit future relapse-associated behavior. Moreover, overexpressing HDAC5 in the NAc suppressed context-associated and reinstated heroin-seeking behaviors, but it did not alter sucrose seeking. We also found that HDAC5 functions within dopamine D1 receptor-expressing MSNs to suppress cue-induced heroin seeking, and within dopamine D2 receptor-expressing MSNs to suppress drug-primed heroin seeking. Assessing cell-type–specific transcriptomics, we found that HDAC5 reduced expression of multiple ion transport genes in both D1- and D2-MSNs. Consistent with this observation, HDAC5 also produced firing rate depression in both MSN classes. These findings revealed roles for HDAC5 during active heroin use in both D1- and D2-MSNs to limit distinct triggers of drug-seeking behavior. Together, our results suggest that HDAC5 might limit relapse vulnerability through regulation of ion channel gene expression and suppression of MSN firing rates during active heroin use.}, number={7}, journal={Proceedings of the National Academy of Sciences}, publisher={Proceedings of the National Academy of Sciences}, author={Anderson, Ethan M. and Tsvetkov, Evgeny and Galante, Allison and DeVries, Derek and McCue, Lauren M. and Wood, Daniel and Barry, Sarah and Berto, Stefano and Lavin, Antonieta and Taniguchi, Makoto and et al.}, year={2023}, month={Feb} } @article{siemsen_barry_vollmer_green_brock_westphal_king_devries_otis_cowan_et al._2022, title={A Subset of Nucleus Accumbens Neurons Receiving Dense and Functional Prelimbic Cortical Input Are Required for Cocaine Seeking}, volume={16}, url={http://dx.doi.org/10.3389/fncel.2022.844243}, DOI={10.3389/fncel.2022.844243}, abstractNote={BackgroundPrelimbic cortical projections to the nucleus accumbens core are critical for cue-induced cocaine seeking, but the identity of the accumbens neuron(s) targeted by this projection, and the transient neuroadaptations contributing to relapse within these cells, remain unknown.}, journal={Frontiers in Cellular Neuroscience}, publisher={Frontiers Media SA}, author={Siemsen, Benjamin M. and Barry, Sarah M. and Vollmer, Kelsey M. and Green, Lisa M. and Brock, Ashley G. and Westphal, Annaka M. and King, Raven A. and DeVries, Derek M. and Otis, James M. and Cowan, Christopher W. and et al.}, year={2022}, month={Feb} }